WO2009105284A1 - Procédés de préparation de composés macrocycliques - Google Patents

Procédés de préparation de composés macrocycliques Download PDF

Info

Publication number
WO2009105284A1
WO2009105284A1 PCT/US2009/001297 US2009001297W WO2009105284A1 WO 2009105284 A1 WO2009105284 A1 WO 2009105284A1 US 2009001297 W US2009001297 W US 2009001297W WO 2009105284 A1 WO2009105284 A1 WO 2009105284A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
compounds
acid
alkyl
mmol
Prior art date
Application number
PCT/US2009/001297
Other languages
English (en)
Inventor
Hendrik Luesch
Jiyong Hong
Original Assignee
University Of Florida Research Foundation, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University Of Florida Research Foundation, Inc. filed Critical University Of Florida Research Foundation, Inc.
Priority to US12/918,958 priority Critical patent/US8759512B2/en
Publication of WO2009105284A1 publication Critical patent/WO2009105284A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains three hetero rings
    • C07D513/16Peri-condensed systems

Definitions

  • the invention is directed towards methods of preparing macrocyclic compounds, such compounds being useful for modulating proliferation activity, and methods of treating proliferation disease and disorders.
  • the methods include those delineated in the schemes and figures herein, including one or any combination of intermediates, chemical transformations or reagents specifically exemplified herein.
  • the invention provides methods of preparing a compound according to Formula I:
  • the method comprises one or more chemical transformations delineated in the Examples herein.
  • Another aspect is a method for preparing a compound of formula Ia (and pharmaceutically acceptable salts, solvates, or hydrates thereof), where R, R 1 , R 2 , R 3 , and R 4 are as defined in formula I:
  • the method comprising one or more chemical transformations delineated in Scheme 2.
  • the method comprises one or more chemical transformations delineated in the Examples herein.
  • R 3 and R 4 are H; wherein R 1 is isopropyl; wherein R 2 is alkyl; wherein R 2 is alkylC(O)-; wherein R 2 is H; wherein the compound is any of Compounds 21-28 in Table A; or wherein the compound is largazole.
  • the chemicals used in the synthetic routes delineated herein may include, for example, solvents, reagents, catalysts, and protecting group and deprotecting group reagents.
  • the methods described above may also additionally include steps, either before or after the steps described specifically herein, to add or remove suitable protecting groups in order to ultimately allow synthesis of the compounds.
  • various synthetic steps may be performed in an alternate sequence or order to give the desired compounds.
  • Synthetic chemistry transformations and protecting group methodologies protecting group methodologies (protection and deprotection) useful in synthesizing applicable compounds are known in the art and include, for example, those described in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T. W. Greene and P. G. M.
  • the invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the compound of formula I and a pharmaceutically acceptable carrier, made by a process delineated herein.
  • the compounds made by the processes herein are useful for modulating the proliferation activity in a subject, by contacting the subject with a compound of formula I, in an amount and under conditions sufficient to modulate proliferation activity.
  • the compounds made by the processes herein are useful for treating a subject suffering from or susceptible to a proliferation related disorder or disease, by administering to the subject an effective amount of a compound or pharmaceutical composition of formula I.
  • the compounds made by the processes herein are useful for treating cancer, tumor growth, cancer of the colon, breast, bone, brain and others (e.g., osteosarcoma, neuroblastoma, colon adenocarcinoma) , by administering to said subject in need thereof, an effective amount of a compound delineated herein (e.g., Formula I), and pharmaceutically acceptable salts thereof.
  • a compound delineated herein e.g., Formula I
  • cardiac cancer e.g., sarcoma, myxoma, rhabdomyoma, fibroma, lipoma and teratoma
  • lung cancer e.g., bronchogenic carcinoma, alveolar carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma
  • various gastrointestinal cancer e.g., cancers of esophagus, stomach, pancreas, small bowel, and large bowel
  • genitourinary tract cancer e.g., kidney, bladder and urethra, prostate, testis
  • liver cancer e.g., hepatoma, cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma
  • bone cancer e.g., osteogenic s
  • hematologic cancer e.g., cancers relating to blood, Hodgkin's disease, non-Hodgkin's lymphoma
  • skin cancer e.g., malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis
  • cancers of the adrenal glands e.g., neuroblastoma.
  • the compounds made by the processes herein are useful for inhibiting histone deacetylase (HDAC) in a subject in need thereof by administering to said subject, an effective amount of a compound delineated herein (e.g., Formula I), and pharmaceutically acceptable salts thereof.
  • HDAC histone deacetylase
  • HDAC histone deacetylase
  • HDAC inhibitors also have widespread modulatory effects on gene expression within the immune system and have been used successfully in the lupus and rheumatoid arthritis autoimmune disease models. Recently, the efficacy of the HDAC inhibitor Trichostatin A was established in ameliorating disease in the multiple sclerosis (MS) animal model, experimental autoimmune encephalomyelitis (EAE).
  • MS multiple sclerosis
  • EAE experimental autoimmune encephalomyelitis
  • the compounds herein are useful to treat MS, an autoimmune, demyelinating and degenerative disease of the human central nervous system (CNS).
  • CNS central nervous system
  • the compounds herein are useful to treat stroke.
  • treating a disorder encompasses preventing, ameliorating, mitigating and/or managing the disorder and/or conditions that may cause the disorder.
  • the terms “treating” and “treatment” refer to a method of alleviating or abating a disease and/or its attendant symptoms.
  • “treating” includes preventing, blocking, inhibiting, attenuating, protecting against, modulating, reversing the effects of and reducing the occurrence of e.g., the harmful effects of a disorder.
  • inhibiting encompasses preventing, reducing and halting progression.
  • modulate refers to increases or decreases in the activity of a cell in response to exposure to a compound of the invention.
  • isolated refers to material that is substantially or essentially free from components that normally accompany it as found in its native state. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. Particularly, in embodiments the compound is at least 85% pure, more preferably at least 90% pure, more preferably at least 95% pure, and most preferably at least 99% pure.
  • polypeptide peptide
  • protein protein
  • amino acid polymers in which one or more amino acid residue is an artificial chemical mimetic of a corresponding naturally occurring amino acid, as well as to naturally occurring amino acid polymers and non-naturally occurring amino acid polymer.
  • a “peptide” is a sequence of at least two amino acids. Peptides can consist of short as well as long amino acid sequences, including proteins.
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, ⁇ -carboxyglutamate, and O-phosphoserine.
  • Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that functions in a manner similar to a naturally occurring amino acid.
  • protein refers to series of amino acid residues connected one to the other by peptide bonds between the alpha- amino and carboxy groups of adjacent residues.
  • Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission.
  • amino acid sequences As to amino acid sequences, one of skill will recognize that individual substitutions, deletions or additions to a peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a "conservatively modified variant" where the alteration results in the substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are well known in the art.
  • Macromolecular structures such as polypeptide structures can be described in terms of various levels of organization. For a general discussion of this organization, see, e.g., Alberts et al., Molecular Biology of the Cell (3rd ed., 1994) and Cantor and Schimmel, Biophysical Chemistry Part I. The Conformation of Biological Macromolecules (1980).
  • Primary structure refers to the amino acid sequence of a particular peptide.
  • Secondary structure refers to locally ordered, three dimensional structures within a polypeptide. These structures are commonly known as domains. Domains are portions of a polypeptide that form a compact unit of the polypeptide and are typically 50 to 350 amino acids long.
  • Typical domains are made up of sections of lesser organization such as stretches of ⁇ -sheet and ⁇ -helices.
  • Tetiary structure refers to the complete three dimensional structure of a polypeptide monomer.
  • Quaternary structure refers to the three dimensional structure formed by the noncovalent association of independent tertiary units. Anisotropic terms are also known as energy terms.
  • administration or “administering” includes routes of introducing the compound(s) to a subject to perform their intended function. Examples of routes of administration which can be used include injection (subcutaneous, intravenous, parenterally, intraperitoneally, intrathecal), topical, oral, inhalation, rectal and transdermal.
  • an effective amount includes an amount effective, at dosages and for periods of time necessary, to achieve the desired result.
  • An effective amount of compound may vary according to factors such as the disease state, age, and weight of the subject, and the ability of the compound to elicit a desired response in the subject. Dosage regimens may be adjusted to provide the optimum therapeutic response.
  • An effective amount is also one in which any toxic or detrimental effects (e.g., side effects) of the elastase inhibitor compound are outweighed by the therapeutically beneficial effects.
  • systemic administration means the administration of a compound(s), drug or other material, such that it enters the patient's system and, thus, is subject to metabolism and other like processes.
  • therapeutically effective amount refers to that amount of the compound being administered sufficient to prevent development of or alleviate to some extent one or more of the symptoms of the condition or disorder being treated.
  • a therapeutically effective amount of compound may range from about 0.005 ⁇ g/kg to about 200 mg/kg, preferably about 0.1 mg/kg to about 200 mg/kg, more preferably about 10 mg/kg to about 100 mg/kg of body weight. In other embodiments, the therapeutically effect amount may range from about 1.0 pM to about 50OnM.
  • treatment of a subject with a therapeutically effective amount of a compound can include a single treatment or, preferably, can include a series of treatments.
  • a subject is treated with a compound in the range of between about 0.005 ⁇ g/kg to about 200 mg/kg of body weight, one time per week for between about 1 to 10 weeks, preferably between 2 to 8 weeks, more preferably between about 3 to 7 weeks, and even more preferably for about 4, 5, or 6 weeks.
  • the effective dosage of a compound used for treatment may increase or decrease over the course of a particular treatment.
  • the term "chiral" refers to molecules which have the property of non- superimposability of the mirror image partner, while the term “achiral” refers to molecules which are superimposable on their mirror image partner.
  • diastereomers refers to stereoisomers with two or more centers of dissymmetry and whose molecules are not mirror images of one another.
  • enantiomers refers to two stereoisomers of a compound which are non-superimposable mirror images of one another.
  • An equimolar mixture of two enantiomers is called a “racemic mixture” or a “racemate.”
  • isomers or “stereoisomers” refers to compounds which have identical chemical constitution, but differ with regard to the arrangement of the atoms or groups in space.
  • prodrug includes compounds with moieties which can be metabolized in vivo. Generally, the prodrugs are metabolized in vivo by esterases or by other mechanisms to active drugs. Examples of prodrugs and their uses are well known in the art (See, e.g., Berge et al. (1977) "Pharmaceutical Salts", J. Pharm. Sci. 66: 1-19).
  • the prodrugs can be prepared in situ during the final isolation and purification of the compounds, or by separately reacting the purified compound in its free acid form or hydroxyl with a suitable esterifying agent. Hydroxyl groups can be converted into esters via treatment with a carboxylic acid.
  • prodrug moieties include substituted and unsubstituted, branch or unbranched lower alkyl ester moieties, (e.g., propionoic acid esters), lower alkenyl esters, di-lower alkyl-amino lower-alkyl esters (e.g., dimethylaminoethyl ester), acylamino lower alkyl esters (e.g., acetyloxymethyl ester), acyloxy lower alkyl esters (e.g., pivaloyloxymethyl ester), aryl esters (phenyl ester), aryl-lower alkyl esters (e.g., benzyl ester), substituted (e.g., with methyl, halo, or methoxy substituents) aryl and aryl-lower alkyl esters, amides, lower-alkyl amides, di-lower alkyl amides, and hydroxy amides.
  • prodrug moieties are propionoic acid esters and acyl esters.
  • Prodrugs which are converted to active forms through other mechanisms in vivo are also included.
  • the compounds of the invention are prodrugs of any of the formulae herein.
  • subject refers to animals such as mammals, including, but not limited to, primates (e.g., humans), cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice and the like. In certain embodiments, the subject is a human.
  • the compounds of the invention include olefins having either geometry: "Z” refers to what is referred to as a “cis” (same side) conformation whereas “E” refers to what is referred to as a “trans” (opposite side) conformation.
  • Z refers to what is referred to as a "cis” (same side) conformation
  • E refers to what is referred to as a "trans” (opposite side) conformation.
  • d and “1" configuration are as defined by the IUPAC Recommendations.
  • diastereomer, racemate, epimer and enantiomer these will be used in their normal context to describe the stereochemistry of preparations.
  • alkyl refers to a straight-chained or branched hydrocarbon group containing 1 to 12 carbon atoms.
  • the term “lower alkyl” refers to a C1-C6 alkyl chain. Examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, tert-butyl, and n-pentyl. Alkyl groups may be optionally substituted with one or more substituents.
  • alkenyl refers to an unsaturated hydrocarbon chain that may be a straight chain or branched chain, containing 2 to 12 carbon atoms and at least one carbon-carbon double bond. Alkenyl groups may be optionally substituted with one or more substituents.
  • alkynyl refers to an unsaturated hydrocarbon chain that may be a straight chain or branched chain, containing the 2 to 12 carbon atoms and at least one carbon-carbon triple bond. Alkynyl groups may be optionally substituted with one or more substituents.
  • the sp or sp carbons of an alkenyl group and an alkynyl group, respectively, may optionally be the point of attachment of the alkenyl or alkynyl groups.
  • alkoxy refers to an -O-alkyl radical.
  • halogen means -F, -Cl, -Br or -I.
  • cycloalkyl refers to a hydrocarbon 3-8 membered monocyclic or 7-14 membered bicyclic ring system having at least one saturated ring or having at least one non-aromatic ring, wherein the non-aromatic ring may have some degree of unsaturation.
  • Cycloalkyl groups may be optionally substituted with one or more substituents. In one embodiment, 0, 1, 2, 3, or 4 atoms of each ring of a cycloalkyl group may be substituted by a substituent.
  • cycloalkyl group examples include cyclopropyl, cyclopentyl, cyclohexyl, cyclobutyl, cycloheptyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, and the like.
  • aryl refers to a hydrocarbon monocyclic, bicyclic or tricyclic aromatic ring system.
  • Aryl groups may be optionally substituted with one or more substituents. In one embodiment, 0, 1, 2, 3, 4, 5 or 6 atoms of each ring of an aryl group may be substituted by a substituent. Examples of aryl groups include phenyl, naphthyl, anthracenyl, fluorenyl, indenyl, azulenyl, and the like.
  • heteroaryl refers to an aromatic 5-8 membered monocyclic, 8-12 membered bicyclic, or 11-14 membered tricyclic ring system having 1-4 ring heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, or S, and the remainder ring atoms being carbon (with appropriate hydrogen atoms unless otherwise indicated).
  • Heteroaryl groups may be optionally substituted with one or more substituents. In one embodiment, 0, 1, 2, 3, or 4 atoms of each ring of a heteroaryl group may be substituted by a substituent.
  • heteroaryl groups include pyridyl, furanyl, thienyl, pyrrolyl, oxazolyl, oxadiazolyl, imidazolyl thiazolyl, isoxazolyl, quinolinyl, pyrazolyl, isothiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, isoquinolinyl, indazolyl, and the like.
  • heterocycloalkyl refers to a nonaromatic 3-8 membered monocyclic, 7-12 membered bicyclic, or 10-14 membered tricyclic ring system comprising 1-3 heteroatoms if monocyclic, 1-6 heteroatoms if bicyclic, or 1-9 heteroatoms if tricyclic, said heteroatoms selected from O, N, S, B, P or Si, wherein the nonaromatic ring system is completely saturated.
  • Heterocycloalkyl groups may be optionally substituted with one or more substituents. In one embodiment, 0, 1, 2, 3, or 4 atoms of each ring of a heterocycloalkyl group may be substituted by a substituent.
  • heterocycloalkyl groups include piperidinyl, piperazinyl, tetrahydropyranyl, morpholinyl, thiomorpholinyl, 1,3-dioxolane, tetrahydrofuranyl, tetrahydrothienyl, thiirenyl, and the like.
  • alkylamino refers to an amino substituent which is further substituted with one or two alkyl groups.
  • aminoalkyl refers to an alkyl substituent which is further substituted with one or more amino groups.
  • hydroxyalkyl or hydroxylalkyl refers to an alkyl substituent which is further substituted with one or more hydroxyl groups.
  • alkyl or aryl portion of alkylamino, aminoalkyl, mercaptoalkyl, hydroxyalkyl, mercaptoalkoxy, sulfonylalkyl, sulfonylaryl, alkylcarbonyl, and alkyl carbonyl alkyl may be optionally substituted with one or more substituents.
  • Acids and bases useful in the methods herein are known in the art.
  • Acid catalysts are any acidic chemical, which can be inorganic (e.g., hydrochloric, sulfuric, nitric acids, aluminum trichloride) or organic (e.g., camphorsulfonic acid, p- toluenesulfonic acid, acetic acid, ytterbium triflate) in nature. Acids are useful in either catalytic or stoichiometric amounts to facilitate chemical reactions.
  • Bases are any basic chemical, which can be inorganic (e.g., sodium bicarbonate, potassium hydroxide) or organic (e.g., triethylamine, pyridine) in nature. Bases are useful in either catalytic or stoichiometric amounts to facilitate chemical reactions.
  • Alkylating agents are any reagent that is capable of effecting the alkylation of the functional group at issue (e.g., oxygen atom of an alcohol, nitrogen atom of an amino group).
  • Alkylating agents are known in the art, including in the references cited herein, and include alkyl halides (e.g., methyl iodide, benzyl bromide or chloride), alkyl sulfates (e.g., methyl sulfate), or other alkyl group-leaving group combinations known in the art.
  • Leaving groups are any stable species that can detach from a molecule during a reaction (e.g., elimination reaction, substitution reaction) and are known in the art, including in the references cited herein, and include halides (e.g., I-, Cl-, Br-, F-), hydroxy, alkoxy (e.g., -OMe, -Ot-Bu), acyloxy anions (e.g., - OAc, -OC(O)CF 3 ), sulfonates (e.g., mesyl, tosyl), acetamides (e.g., -NHC(O)Me), carbamates (e.g., N(Me)C(O)Ot-Bu), phosphonates (e.g., -OP(O)(OEt) 2 ), water or alcohols (protic conditions), and the like.
  • halides e.g., I-, Cl-, Br-, F-
  • alkoxy
  • substituents on any group can be at any atom of that group, wherein any group that can be substituted (such as, for example, alkyl, alkenyl, alkynyl, aryl, aralkyl, heteroaryl, heteroaralkyl, cycloalkyl, heterocycloalkyl) can be optionally substituted with one or more substituents (which may be the same or different), each replacing a hydrogen atom.
  • substituents include, but are not limited to alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aralkyl, heteroaralkyl, aryl, heteroaryl, halogen, haloalkyl, cyano, nitro, alkoxy, aryloxy, hydroxyl, hydroxyl alkyl, oxo (i.e., carbonyl), carboxyl, formyl, alkylcarbonyl, alkylcarbonylalkyl, alkoxycarbonyl, alkylcarbonyloxy, aryloxycarbonyl, heteroaryloxy, heteroaryloxycarbonyl, thio, mercapto, mercaptoalkyl, arylsulfonyl, amino, aminoalkyl, dialkylamino, alkylcarbonylamino, alkylaminocarbonyl, alkoxycarbonylamino, alkylamino, arylamino, diary
  • the putative glycine carbonyl ( ⁇ c-n 167.9) was part of a 2,4-disubstituted thiazole unit as evidenced by HMBCs from the only aromatic methine (6 H - I2 7.76, ⁇ c-i 2 124.2) to C-13 and to another quaternary sp 2 carbon, C-11 ( ⁇ c 147.4).
  • the only other HMBC to C-IO was from the thiazole proton H- 12, indicating that C-IO bore the thiazole substituent.
  • This unit was attached to the amino terminus of the glycine-derived unit as shown by HMBCs from 14-NH and H-14a/b to C-15 as well as ROESY cross peaks between 14-NH and H- 16a and H-16b.
  • the last unit was an n-octanoyl group (C-22 to C-29) which was connected with C-21 based on HMBC from H 2 -21 to C-22.
  • the low-field chemical shift for C-22 ( ⁇ c 199.4) coupled with the fact that one sulfur atom yet remained to be assigned was strong evidence for a thioester functionality.
  • Largazole (1) possesses a dense combination of unusual structural features, including a substituted 4-methylthiazoline linearly fused to a thiazole, previously only found in didehydromirabazole, 7 a member of the group of terrestrial cyanobacterial cytotoxins from Scytonema mirabile with solid tumor selectivity. Another remarkable structural element is the thioester moiety; thioester-containing secondary metabolites have been reported previously from sponges, 9 eukaryotic algae 10 and bacteria," but not from cyanobacteria. The 3-hydroxy-7-thio-hept-4-enoic acid unit in 1 is unprecedented in natural products.
  • Beilstein® (Elsevier MDL), or by appropriate keyword searching using an internet search engine such as Google® or keyword databases such as the US Patent and Trademark Office text database.
  • the compounds herein may also contain linkages (e.g., carbon-carbon bonds) wherein bond rotation is restricted about that particular linkage, e.g. restriction resulting from the presence of a ring or double bond. Accordingly, all cis/trans and E/Z isomers are expressly included in the present invention.
  • the compounds herein may also be represented in multiple tautomeric forms, in such instances, the invention expressly includes all tautomeric forms of the compounds described herein, even though only a single tautomeric form may be represented. All such isomeric forms of such compounds herein are expressly included in the present invention. All crystal forms and polymorphs of the compounds described herein are expressly included in the present invention. Also embodied are extracts and fractions comprising compounds of the invention.
  • isomers is intended to include diastereoisomers, enantiomers, regioisomers, structural isomers, rotational isomers, tautomers, and the like.
  • the methods of the invention may be carried out with an enantiomerically enriched compound, a racemate, or a mixture of diastereomers.
  • Preferred enantiomerically enriched compounds have an enantiomeric excess of 50% or more, more preferably the compound has an enantiomeric excess of 60%, 70%, 80%, 90%, 95%, 98%, or 99% or more.
  • only one enantiomer or diastereomer of a chiral compound of the invention is administered to cells or a subject.
  • Another object of the present invention is the use of a compound as described herein (e.g., of any formulae herein) in the manufacture of a medicament for use in the treatment of a cell proliferation disorder or disease.
  • Another object of the present invention is the use of a compound as described herein (e.g., of any formulae herein) for use in the treatment of a cell proliferation disorder or disease.
  • compositions in one aspect, the invention provides a pharmaceutical composition comprising the compound of formula I made by the processes herein and a pharmaceutically acceptable carrier.
  • the invention provides a pharmaceutical composition wherein the compound of formula I is largazole made by the processes herein, and a pharmaceutically acceptable carrier.
  • the invention provides a pharmaceutical composition further comprising an additional therapeutic agent.
  • the additional therapeutic agent is an anti-cancer agent, chemotherapeutic agent, an anti- angiogenesis agent, cytotoxic agent, or an anti-proliferation agent.
  • pharmaceutically acceptable salts or “pharmaceutically acceptable carrier” is meant to include salts of the active compounds which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein.
  • base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent.
  • pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt.
  • acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent.
  • Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p- tolylsulfonic, citric, tartaric, methanesulfonic, and the like.
  • inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and
  • salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, e.g., Berge et al., Journal of Pharmaceutical Science 66:1-19 (1977)).
  • Certain specific compounds of the present invention contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
  • Other pharmaceutically acceptable carriers known to those of skill in the art are suitable for the present invention.
  • the neutral forms of the compounds may be regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner.
  • the parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present invention.
  • the present invention provides compounds which are in a prodrug form.
  • Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present invention.
  • prodrugs can be converted to the compounds of the present invention by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to the compounds of the present invention when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent.
  • Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are intended to be encompassed within the scope of the present invention. Certain compounds of the present invention may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.
  • Actual dosage levels and time course of administration of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic (or unacceptably toxic) to the patient.
  • At least one compound according to the present invention is administered in a pharmaceutically effective amount to a subject in need thereof in a pharmaceutical carrier by intravenous, intramuscular, subcutaneous, or intracerebro ventricular injection or by oral administration or topical application.
  • a compound of the invention may be administered alone or in conjunction with a second, different therapeutic.
  • in conjunction with is meant together, substantially simultaneously or sequentially.
  • a compound of the invention is administered acutely.
  • the compound of the invention may therefore be administered for a short course of treatment, such as for about 1 day to about 1 week.
  • the compound of the invention may be administered over a longer period of time to ameliorate chronic disorders, such as, for example, for about one week to several months depending upon the condition to be treated.
  • pharmaceutically effective amount as used herein is meant an amount of a compound of the invention, high enough to significantly positively modify the condition to be treated but low enough to avoid serious side effects (at a reasonable benefit/risk ratio), within the scope of sound medical judgment.
  • a pharmaceutically effective amount of a compound of the invention will vary with the particular goal to be achieved, the age and physical condition of the patient being treated, the severity of the underlying disease, the duration of treatment, the nature of concurrent therapy and the specific organozinc compound employed. For example, a therapeutically effective amount of a compound of the invention administered to a child or a neonate will be reduced proportionately in accordance with sound medical judgment. The effective amount of a compound of the invention will thus be the minimum amount which will provide the desired effect.
  • a decided practical advantage of the present invention is that the compound may be administered in a convenient manner such as by intravenous, intramuscular, subcutaneous, oral or intra-cerebro ventricular injection routes or by topical application, such as in creams or gels.
  • the active ingredients which comprise a compound of the invention may be required to be coated in a material to protect the compound from the action of enzymes, acids and other natural conditions which may inactivate the compound.
  • the compound can be coated by, or administered with, a material to prevent inactivation.
  • Dispersions can also be prepared, for example, in glycerol, liquid polyethylene glycols, and mixtures thereof, and in oils.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the carrier can be a solvent or dispersion medium containing, for example, water, DMSO, ethanol, polyol (for example, glycerol, propylene glycol, liquid polyethylene glycol, and the like), suitable mixtures thereof and vegetable oils.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion.
  • a coating such as lecithin
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the compound of the invention in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the various sterilized compounds into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and the freeze-drying technique which yields a powder of the active ingredient plus any additional desired ingredient from previously sterile-filtered solution thereof.
  • the compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • compositions or preparations according to the present invention are prepared so that an oral dosage unit form contains compound concentration sufficient to treat a disorder in a subject.
  • substances which can serve as pharmaceutical carriers are sugars, such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethycellulose, ethylcellulose and cellulose acetates; powdered tragancanth; malt; gelatin; talc; stearic acids; magnesium stearate; calcium sulfate; vegetable oils, such as peanut oils, cotton seed oil, sesame oil, olive oil, corn oil and oil of theobroma; polyols such as propylene glycol, glycerine, sorbitol, manitol, and polyethylene glycol; agar; alginic acids; pyrogen- free water; isotonic saline; and phosphate buffer solution; skim milk powder; as well as other non-toxic compatible substances used in
  • HRMS data were obtained using an Agilent LC-TOF mass spectrometer equipped with an ESI/ APCI multimode ion source detector.
  • Enantiomeric standards of 2-methylcysteic acid were obtained by oxidation of (R)- and (5)-2-methylcysteines (see below) which were provided by ResCom (DSM Pharma Chemicals).
  • Valine, glycine and malic acid standards were obtained from Sigma.
  • Paclitaxel, actinomycin D and doxorubicin were obtained from EMD Chemicals, Inc.
  • Symploca sp. was collected from Pillars, Key Largo (Florida Keys, USA) in August 2003. The specimens had upright, golden-brown, feather-like filaments consistent with this genus. Filaments measured 5-6 ⁇ m in width including a fine sheath and 8-9 ⁇ m in length. Symploca sp. was freeze-dried and extracted with MeOH-EtOAc (1 :1). The resulting lipophilic extract (0.29 g) was partitioned between hexanes and 20% aqueous MeOH. The aqueous MeOH layer was concentrated and fractionated by Si gel chromatography using CH 2 Cl 2 containing increasing amounts of z-PrOH followed by MeOH.
  • Example 3 Determination of Absolute Configuration.
  • a sample of compound 21 ( ⁇ 100 ⁇ g) was dissolved in 4 mL Of CH 2 Cl 2 and subjected to ozonolysis at room temperature for 30 min. The solvent was evaporated and the residue was treated with 0.6 mL Of H 2 O 2 -HCO 2 H (1 :2) at 70 0 C for 20 min. The solvent was evaporated and the resulting oxidation product was hydrolyzed with 0.5 mL of 6 N HCl at 1 10 0 C for 24 h.
  • the hydrolyzed product was dried and analyzed by chiral HPLC (column, Phenomenex Chirex phase 3126 N.S-dioctyl- ⁇ )- pencillamine, 4.60 x 250 mm, 5 ⁇ m; solvent 1, 2 mM CuSO 4 in 95:5 H 2 O/MeC ⁇ , pH 4.50; solvent 2, 0.5 mM Cu(OAc) 2 /0.1 M NH 4 OAc in 85:15 H 2 O/MeCN, pH 4.6; flow rate 1.0 mL/min; detection at 254 nm).
  • the absolute configuration of the amino acids in the hydrolyzate was determined by direct comparison with the retention times of authentic standards.
  • the retention times (t R , min) for solvent 1 were as follows: GIy (5.3), L-VaI (12.6), D-VaI (16.4), (S)-2-Me-cysteic acid (20.0), and ( ⁇ )-2-Me- cysteic acid (23.9).
  • the retention times (J R , min) of the hydrolyzate components were 5.3, 12.6, 23.9, indicating the presence of GIy, L-VaI and (/?)-2-Me-cysteic acid in the product mixture.
  • Solvent 2 was used to detect malic acid. Standard L-malic acid eluted at f ⁇ 7.6 min and D-malic acid at t R 20.4 min.
  • Cell culture medium was purchased from Invitrogen and fetal bovine serum (FBS) from Hyclone. Cells were propagated and maintained in DMEM medium (high glucose) supplemented with 10% FBS at 37 0 C humidified air and 5% CO 2 .
  • FBS fetal bovine serum
  • Cells suspended in DMEM containing 10% FBS were plated in 96-well plates (MDA-MB-231 : 12,000 cells; NMuMG: 5,000 cells; U2OS: 5,000 cells; HT29: 10,000 cells; IMR-32: 30,000 cells; NIH3T3: 5,000 cells) incubated (37 °C, 5% CO 2 ) and 24 h later treated with various concentrations of compound 21 or solvent control (1% EtOH). After another 48 h of incubation, cell viability was measured using MTT according to manufacturer's instructions (Promega).
  • Example 6 Anticancer therapeutics activity MDA-MB-231 and NMuMG cells were also treated with paclitaxel (in
  • GI 50 and LCs 0 values were calculated as previously described (K. D. Paull, E. Hamel, L. Malspeis, In Cancer C 'hemo therapeutic Agents, W. E. Foye, Ed., American Chemical Society, Washington, DC, 1995, pp. 10-1 1).
  • Largazole (21) potently inhibited the growth of highly invasive transformed human mammary epithelial cells (MDA-MB-231) in a dose-dependent manner (GI 50 8 nM) and induced cytotoxicity at higher concentrations (LC 50 117 nM) based on MTT assay.
  • MDA-MB-231 highly invasive transformed human mammary epithelial cells
  • LC 50 117 nM induced cytotoxicity at higher concentrations
  • NMuMG nontransformed murine mammary epithelial cells
  • Largazole (21) also demonstrates remarkable selectivity that is not observed with other validated antitumor natural products tested in parallel. See, e.g., Table 2 re MDA-MB-231 /NmuMG cells and U2OS/NIH3T3 cells.
  • Table 2 Growth-inhibitory activity (GI 50 ) of natural product drugs
  • Largazole (21) inhibited HDAC activity from a HeLa cell nuclear protein extract rich in class I HDACs 1, 2, and 3 (BIOMOL); however, it is possible that the thioester is cleaved under assay conditions.
  • thiol 29 is a reactive species, we liberated 29 from the acetyl analog 28 of largazole (21) and measured enzymatic activity directly; thiol 9 inhibited the HDACs in the nuclear extract of HeLa cells with a similar IC 5O value (Table 3).
  • Largazole (21) and thiol 29 exhibited similar cellular activity against HDACs derived from nuclear HeLa extracts as well as antiproliferative activity.
  • HCT-116 HCT-116 HeLa growth HDAC nuclear inhibition cellular assay extract
  • the carboxylic acid 16 was treated with TFA/CH 2 C1 2 (1 :5, 5 mL). After being stirred at room temperature for 2 h, the residue was purged by N 2 to remove TFA and CH 2 Cl 2 , then washed with Et 2 O four times to remove remaining TFA. To a solution of the crude 7 in CH 2 Cl 2 (325 mL, 1 mM) was added HATU (247 mg, 0.65 mmol), HOAt (88 mg, 0.65 mmol), and i- Pr 2 NEt (0.24 mL, 1.30 mmol). The reaction mixture was stirred at room temperature for 24 h and then concentrated in vacuo. The residue was diluted with H 2 O and
  • thioester 2 To a solution of thioester 2 (2.3 mg, 0.0043 mmol) in CH 3 CN (0.5 mL, 0.0086 M) was added aqueous NH 3 (28.9%, 0.05 mL). The resulting mixture was stirred at room temperature for 12 h and concentrated in vacuo. The residue was purified by column chromatography (silica gel, EtOAc/MeOH, 10/0 to 10/1) to afford thiol 3 as a colorless oil (1.7 mg, 80%).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention porte sur des procédés de production de composés macrocycliques ayant une activité anti-prolifération, et utiles dans des procédés de traitement de troubles tels que le cancer, de tumeurs et de troubles liés à la prolifération cellulaire.
PCT/US2009/001297 2008-02-24 2009-02-24 Procédés de préparation de composés macrocycliques WO2009105284A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/918,958 US8759512B2 (en) 2008-02-24 2009-02-24 Methods of preparation of macrocyclic compounds

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US3099608P 2008-02-24 2008-02-24
US61/030,996 2008-02-24

Publications (1)

Publication Number Publication Date
WO2009105284A1 true WO2009105284A1 (fr) 2009-08-27

Family

ID=40985859

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/001297 WO2009105284A1 (fr) 2008-02-24 2009-02-24 Procédés de préparation de composés macrocycliques

Country Status (2)

Country Link
US (1) US8759512B2 (fr)
WO (1) WO2009105284A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2276757A2 (fr) * 2008-04-11 2011-01-26 University of Florida Research Foundation, Inc. Composés macrocycliques et procédés de traitement
EP2574194A2 (fr) * 2010-05-21 2013-04-03 University of Florida Research Foundation, Inc. Composés macrocycliques et méthodes de traitement
WO2015200699A3 (fr) * 2014-06-26 2016-05-26 University Of Florida Research Foundation Composés macrocycliques et méthodes de traitement associées

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9492434B2 (en) 2011-12-09 2016-11-15 University Of Florida Research Foundation, Inc. Combination compositions and methods of treatment
CN106456613B (zh) * 2014-05-27 2019-07-09 恩库勒公司 制备环缩肽的方法

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
CHEN ET AL.: "Total Synthesis of Apratoxin A.", JOURNAL OF AMERICAN CHEMICAL SOCIETY, vol. 125, 2003, pages 8734 - 8735 *
KASPEREK ET AL.: "Intramolecular Catalysis in the Aminolysis of Thiol Esters.", JOURNAL OF ORGANIC CHEMISTRY, vol. 37, 1972, pages 1456 - 1457 *
TAORI ET AL.: "Structure and Activity of Largazole, a Potent Antiproliferative Agent from the Floridian Marine Cyanobacterium Symploca sp.", JOURNAL OF AMERICA CHEMICAL SOCIETY, vol. 130, 19 January 2008 (2008-01-19), pages 1806 - 1807 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2276757A2 (fr) * 2008-04-11 2011-01-26 University of Florida Research Foundation, Inc. Composés macrocycliques et procédés de traitement
EP2276757A4 (fr) * 2008-04-11 2012-03-28 Univ Florida Composés macrocycliques et procédés de traitement
AU2009234398B2 (en) * 2008-04-11 2015-01-22 University Of Florida Research Foundation, Inc. Macrocyclic compounds and methods of treatment
EP2574194A2 (fr) * 2010-05-21 2013-04-03 University of Florida Research Foundation, Inc. Composés macrocycliques et méthodes de traitement
JP2013526587A (ja) * 2010-05-21 2013-06-24 ユニバーシティ オブ フロリダ リサーチ ファウンデーション,インコーポレイテッド 大環状化合物および治療方法
EP2574194A4 (fr) * 2010-05-21 2014-01-15 Univ Florida Composés macrocycliques et méthodes de traitement
WO2015200699A3 (fr) * 2014-06-26 2016-05-26 University Of Florida Research Foundation Composés macrocycliques et méthodes de traitement associées
US9815852B2 (en) 2014-06-26 2017-11-14 University Of Florida Research Foundation, Incorporated Macrocyclic compounds and methods of treatment

Also Published As

Publication number Publication date
US20110092697A1 (en) 2011-04-21
US8759512B2 (en) 2014-06-24

Similar Documents

Publication Publication Date Title
US9023875B2 (en) Macrocyclic compounds and methods of treatment
US11718645B2 (en) Macrocyclic therapeutic agents, methods of manufacture, and methods of treatment
US20150210718A1 (en) Macrocyclic compounds and methods of treatment
US8759512B2 (en) Methods of preparation of macrocyclic compounds
AU2012255251B2 (en) Macrocyclic therapeutic agents and methods of treatment
US20110294720A1 (en) Apratoxin therapeutic agents: mechanism and methods of treatment
WO2009032338A1 (fr) Agents thérapeutiques à base d'apratoxine : mécanisme et procédés de traitement
US9051350B2 (en) Macrocyclic antiproliferation agents and methods of treatment
WO2009032351A1 (fr) Produits naturels actifs, dérivés, et procédés thérapeutiques
US9815852B2 (en) Macrocyclic compounds and methods of treatment

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 09713124

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 12918958

Country of ref document: US

122 Ep: pct application non-entry in european phase

Ref document number: 09713124

Country of ref document: EP

Kind code of ref document: A1